Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 144(4): 551-65, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21335237

RESUMO

Kinesin and dynein are opposite-polarity microtubule motors that drive the tightly regulated transport of a variety of cargoes. Both motors can bind to cargo, but their overall composition on axonal vesicles and whether this composition directly modulates transport activity are unknown. Here we characterize the intracellular transport and steady-state motor subunit composition of mammalian prion protein (PrP(C)) vesicles. We identify Kinesin-1 and cytoplasmic dynein as major PrP(C) vesicle motor complexes and show that their activities are tightly coupled. Regulation of normal retrograde transport by Kinesin-1 is independent of dynein-vesicle attachment and requires the vesicle association of a complete Kinesin-1 heavy and light chain holoenzyme. Furthermore, motor subunits remain stably associated with stationary as well as with moving vesicles. Our data suggest a coordination model wherein PrP(C) vesicles maintain a stable population of associated motors whose activity is modulated by regulatory factors instead of by structural changes to motor-cargo associations.


Assuntos
Axônios/metabolismo , Dineínas/metabolismo , Cinesinas/metabolismo , Proteínas PrPC/metabolismo , Animais , Hipocampo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora , Neurônios/metabolismo , Vesículas Transportadoras/metabolismo
2.
Hum Mol Genet ; 28(9): 1498-1514, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30590647

RESUMO

Mutations in the microtubule-associated protein tau (MAPT) underlie multiple neurodegenerative disorders, yet the pathophysiological mechanisms are unclear. A novel variant in MAPT resulting in an alanine to threonine substitution at position 152 (A152T tau) has recently been described as a significant risk factor for both frontotemporal lobar degeneration and Alzheimer's disease. Here we use complementary computational, biochemical, molecular, genetic and imaging approaches in Caenorhabditis elegans and mouse models to interrogate the effects of the A152T variant on tau function. In silico analysis suggests that a threonine at position 152 of tau confers a new phosphorylation site. This finding is borne out by mass spectrometric survey of A152T tau phosphorylation in C. elegans and mouse. Optical pulse-chase experiments of Dendra2-tau demonstrate that A152T tau and phosphomimetic A152E tau exhibit increased diffusion kinetics and the ability to traverse across the axon initial segment more efficiently than wild-type (WT) tau. A C. elegans model of tauopathy reveals that A152T and A152E tau confer patterns of developmental toxicity distinct from WT tau, likely due to differential effects on retrograde axonal transport. These data support a role for phosphorylation of the variant threonine in A152T tau toxicity and suggest a mechanism involving impaired retrograde axonal transport contributing to human neurodegenerative disease.


Assuntos
Alelos , Substituição de Aminoácidos , Variação Genética , Proteínas tau/genética , Proteínas tau/metabolismo , Animais , Animais Geneticamente Modificados , Transporte Axonal , Axônios/metabolismo , Caenorhabditis elegans , Modelos Animais de Doenças , Suscetibilidade a Doenças , Humanos , Camundongos , Mutação , Fosforilação , Ligação Proteica , Vesículas Sinápticas/metabolismo , Tauopatias/etiologia , Tauopatias/metabolismo , Tauopatias/patologia
3.
Proc Natl Acad Sci U S A ; 115(33): E7710-E7719, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30061394

RESUMO

Cell-autonomous and cell-nonautonomous mechanisms of neurodegeneration appear to occur in the proteinopathies, including Alzheimer's and Parkinson's diseases. However, how neuronal toxicity is generated from misfolding-prone proteins secreted by nonneuronal tissues and whether modulating protein aggregate levels at distal locales affects the degeneration of postmitotic neurons remains unknown. We generated and characterized animal models of the transthyretin (TTR) amyloidoses that faithfully recapitulate cell-nonautonomous neuronal proteotoxicity by expressing human TTR in the Caenorhabditis elegans muscle. We identified sensory neurons with affected morphological and behavioral nociception-sensing impairments. Nonnative TTR oligomer load and neurotoxicity increased following inhibition of TTR degradation in distal macrophage-like nonaffected cells. Moreover, reducing TTR levels by RNAi or by kinetically stabilizing natively folded TTR pharmacologically decreased TTR aggregate load and attenuated neuronal dysfunction. These findings reveal a critical role for in trans modulation of aggregation-prone degradation that directly affects postmitotic tissue degeneration observed in the proteinopathies.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Pré-Albumina/metabolismo , Agregados Proteicos , Neuropatias Amiloides/genética , Neuropatias Amiloides/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/citologia , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Humanos , Pré-Albumina/genética , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo
4.
Traffic ; 18(1): 71-88, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27770501

RESUMO

In axons, proper localization of proteins, vesicles, organelles, and other cargoes is accomplished by the highly regulated coordination of kinesins and dyneins, molecular motors that bind to cargoes and translocate them along microtubule (MT) tracks. Impairment of axonal transport is implicated in the pathogenesis of multiple neurodegenerative disorders including Alzheimer's and Huntington's diseases. To understand how MT-based cargo motility is regulated and to delineate its role in neurodegeneration, it is critical to analyze the detailed dynamics of moving cargoes inside axons. Here, we present KymoAnalyzer, a software tool that facilitates the robust analysis of axonal transport from time-lapse live-imaging sequences. KymoAnalyzer is an open-source software that automatically classifies particle trajectories and systematically calculates velocities, run lengths, pauses, and a wealth of other parameters that are characteristic of motor-based transport. We anticipate that laboratories will easily use this package to unveil previously uncovered intracellular transport details of individually-moving cargoes inside neurons.


Assuntos
Neurônios/metabolismo , Neurônios/fisiologia , Animais , Transporte Axonal/fisiologia , Axônios/metabolismo , Axônios/fisiologia , Dineínas/metabolismo , Cinesinas/metabolismo , Microtúbulos/metabolismo , Microtúbulos/fisiologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/fisiopatologia , Organelas/metabolismo , Organelas/fisiologia , Software
5.
J Cell Sci ; 127(Pt 7): 1537-49, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24522182

RESUMO

Protein degradation by the ubiquitin-proteasome system in neurons depends on the correct delivery of the proteasome complex. In neurodegenerative diseases, aggregation and accumulation of proteins in axons link transport defects with degradation impairments; however, the transport properties of proteasomes remain unknown. Here, using in vivo experiments, we reveal the fast anterograde transport of assembled and functional 26S proteasome complexes. A high-resolution tracking system to follow fluorescent proteasomes revealed three types of motion: actively driven proteasome axonal transport, diffusive behavior in a viscoelastic axonema and proteasome-confined motion. We show that active proteasome transport depends on motor function because knockdown of the KIF5B motor subunit resulted in impairment of the anterograde proteasome flux and the density of segmental velocities. Finally, we reveal that neuronal proteasomes interact with intracellular membranes and identify the coordinated transport of fluorescent proteasomes with synaptic precursor vesicles, Golgi-derived vesicles, lysosomes and mitochondria. Taken together, our results reveal fast axonal transport as a new mechanism of proteasome delivery that depends on membrane cargo 'hitch-hiking' and the function of molecular motors. We further hypothesize that defects in proteasome transport could promote abnormal protein clearance in neurodegenerative diseases.


Assuntos
Transporte Axonal/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Vesículas Sinápticas/metabolismo , Animais , Axônios/metabolismo , Transporte Biológico , Células Cultivadas , Hipocampo/citologia , Membranas Intracelulares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nervo Isquiático/citologia , Sinaptossomos/metabolismo
6.
J Am Chem Soc ; 137(23): 7404-14, 2015 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-26051248

RESUMO

Fluorogenic probes, due to their often greater spatial and temporal sensitivity in comparison to permanently fluorescent small molecules, represent powerful tools to study protein localization and function in the context of living systems. Herein, we report fluorogenic probe 4, a 1,3,4-oxadiazole designed to bind selectively to transthyretin (TTR). Probe 4 comprises a fluorosulfate group not previously used in an environment-sensitive fluorophore. The fluorosulfate functional group does not react covalently with TTR on the time scale required for cellular imaging, but does red shift the emission maximum of probe 4 in comparison to its nonfluorosulfated analogue. We demonstrate that probe 4 is dark in aqueous buffers, whereas the TTR·4 complex exhibits a fluorescence emission maximum at 481 nm. The addition of probe 4 to living HEK293T cells allows efficient binding to and imaging of exogenous TTR within intracellular organelles, including the mitochondria and the endoplasmic reticulum. Furthermore, live Caenorhabditis elegans expressing human TTR transgenically and treated with probe 4 display TTR·4 fluorescence in macrophage-like coelomocytes. An analogue of fluorosulfate probe 4 does react selectively with TTR without labeling the remainder of the cellular proteome. Studies on this analogue suggest that certain aryl fluorosulfates, due to their cell and organelle permeability and activatable reactivity, could be considered for the development of protein-selective covalent probes.


Assuntos
Caenorhabditis elegans/citologia , Caenorhabditis elegans/metabolismo , Corantes Fluorescentes/química , Fluoretos/química , Organelas/metabolismo , Pré-Albumina/análise , Ácidos Sulfúricos/química , Animais , Sobrevivência Celular , Células Cultivadas , Células HEK293 , Humanos , Modelos Moleculares , Estrutura Molecular , Pré-Albumina/biossíntese , Pré-Albumina/química
7.
Proc Natl Acad Sci U S A ; 109(22): 8582-7, 2012 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-22582169

RESUMO

Intracellular transport of vesicles and organelles along microtubules is powered by kinesin and cytoplasmic dynein molecular motors. Both motors can attach to the same cargo and thus must be coordinated to ensure proper distribution of intracellular materials. Although a number of hypotheses have been proposed to explain how these motors are coordinated, considerable uncertainty remains, in part because of the absence of methods for assessing motor subunit composition on individual vesicular cargos. We developed a robust quantitative immunofluorescence method based on subpixel colocalization to elucidate relative kinesin-1 and cytoplasmic dynein motor subunit composition of individual, endogenous amyloid precursor protein (APP) vesicles in mouse hippocampal cells. The resulting method and data allow us to test a key in vivo prediction of the hypothesis that APP can recruit kinesin-1 to APP vesicles in neuronal axons. We found that APP levels are well-correlated with the amount of the light chain of kinesin-1 (KLC1) and the heavy chain of cytoplasmic dynein (DHC1) on vesicles. In addition, genetic reduction of APP diminishes KLC1 and DHC1 levels on APP cargos. Finally, our data reveal that reduction of KLC1 leads to decreased levels of DHC1 on APP vesicles, suggesting that KLC1 is necessary for the association of DHC1 to these cargos, and help to explain previously reported retrograde transport defects generated when kinesin-1 is reduced.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Dineínas do Citoplasma/metabolismo , Vesículas Citoplasmáticas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Animais Recém-Nascidos , Axônios/metabolismo , Células Cultivadas , Dineínas do Citoplasma/genética , Feminino , Imunofluorescência , Hipocampo/citologia , Hipocampo/metabolismo , Cinesinas , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/genética , Neurônios/metabolismo , Interferência de RNA
8.
Bio Protoc ; 13(7): e4651, 2023 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-37056248

RESUMO

The trafficking and sorting of proteins through the secretory-endolysosomal system is critical for the proper functioning of neurons. Defects in steps of these pathways are associated with neuronal toxicity in various neurodegenerative disorders. The prion protein (PrP) is a glycosylphosphatidylinositol (GPI)-anchored protein that follows the secretory pathway before reaching the cell surface. Following endocytosis from the cell surface, PrP sorts into endosomes and lysosomes for further recycling and degradation, respectively. A few detailed protocols using drug treatments and fluorescent dyes have previously allowed the tracking of PrP trafficking routes in real time in non-neuronal cells. Here, we present a protocol optimized for primary neurons that aims to monitor and/or manipulate the trafficking and sorting of PrP particles at several steps during their secretory-endolysosomal itineraries, including (a) ER export, (b) endocytosis, (c) lysosomal degradation, and (d) accumulation in axonal endolysosomes. These primary neuron live assays allow for the robust quantitation of accumulation and/or degradation of PrP or of other membrane-associated proteins that transition from the ER to the Golgi via the cell surface. Graphical abstract.

9.
bioRxiv ; 2023 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-36993610

RESUMO

Dystrophic axons comprising misfolded mutant prion protein (PrP) aggregates are a characteristic pathological feature in the prionopathies. These aggregates form inside endolysosomes -called endoggresomes-, within swellings that line up the length of axons of degenerating neurons. The pathways impaired by endoggresomes that result in failed axonal and consequently neuronal health, remain undefined. Here, we dissect the local subcellular impairments that occur within individual mutant PrP endoggresome swelling sites in axons. Quantitative high-resolution light and electron microscopy revealed the selective impairment of the acetylated vs tyrosinated microtubule cytoskeleton, while micro-domain image analysis of live organelle dynamics within swelling sites revealed deficits uniquely to the MT-based active transport system that translocates mitochondria and endosomes toward the synapse. Cytoskeletal and defective transport results in the retention of mitochondria, endosomes, and molecular motors at swelling sites, enhancing mitochondria-Rab7 late endosome contacts that induce mitochondrial fission via the activity of Rab7, and render mitochondria dysfunctional. Our findings point to mutant Pr Pendoggresome swelling sites as selective hubs of cytoskeletal deficits and organelle retention that drive the remodeling of organelles along axons. We propose that the dysfunction imparted locally within these axonal micro-domains spreads throughout the axon over time, leading to axonal dysfunction in prionopathies.

10.
Autophagy ; 17(10): 3266-3268, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34424150

RESUMO

Macroautophagy/autophagy is a multi-step process that leads to cargo degradation via the fusion of hydrolases-containing lysosomes with cargo-loaded autophagosomes. For this process to occur, autophagosomes are directionally transported by molecular motors toward the nucleus, where they fuse with lysosomes for cargo degradation. The molecular basis for this regulation, including the cell machinery required for this directional transport, has not been fully identified. Using a combination of proteomic and live-imaging approaches in mammalian cells, including primary neurons, we describe that the phosphorylation of the autophagosome protein Atg8/LC3B by the Hippo kinase STK4/MST1, an event we previously reported to be required for autophagy completion, reduces the binding of the transport-related protein FYCO1 to MAP1LC3B/LC3B. This event in turn allows the proficient microtubule-based transport of autophagosomes toward the perinuclear area, thus facilitating the contact of autophagosomes with lysosomes. In the absence of LC3B phosphorylation, autophagosomes undergo aberrant transport including increased movement toward the cell periphery resulting in reduced autophagosome-lysosome colocalization. Thus, LC3B phosphorylation modulates the directional transport of autophagosomes to meet with lysosomes in the perinuclear area, a crucial event in ensuring autophagic degradation of cargo.


Assuntos
Autofagossomos , Autofagia , Animais , Autofagossomos/metabolismo , Autofagia/fisiologia , Núcleo Celular/metabolismo , Lisossomos/metabolismo , Mamíferos , Fosforilação , Proteômica
11.
Viruses ; 13(7)2021 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-34372599

RESUMO

In prion diseases, the spread of infectious prions (PrPSc) is thought to occur within nerves and across synapses of the central nervous system (CNS). However, the mechanisms by which PrPSc moves within axons and across nerve synapses remain undetermined. Molecular motors, including kinesins and dyneins, transport many types of intracellular cargo. Kinesin-1C (KIF5C) has been shown to transport vesicles carrying the normal prion protein (PrPC) within axons, but whether KIF5C is involved in PrPSc axonal transport is unknown. The current study tested whether stereotactic inoculation in the striatum of KIF5C knock-out mice (Kif5c-/-) with 0.5 µL volumes of mouse-adapted scrapie strains 22 L or ME7 would result in an altered rate of prion spreading and/or disease timing. Groups of mice injected with each strain were euthanized at either pre-clinical time points or following the development of prion disease. Immunohistochemistry for PrP was performed on brain sections and PrPSc distribution and tempo of spread were compared between mouse strains. In these experiments, no differences in PrPSc spread, distribution or survival times were observed between C57BL/6 and Kif5c-/- mice.


Assuntos
Encéfalo/virologia , Cinesinas/genética , Doenças Priônicas/fisiopatologia , Príons/patogenicidade , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
Sci Adv ; 7(52): eabg3693, 2021 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-34936461

RESUMO

The pathogenic aggregation of misfolded prion protein (PrP) in axons underlies prion disease pathologies. The molecular mechanisms driving axonal misfolded PrP aggregate formation leading to neurotoxicity are unknown. We found that the small endolysosomal guanosine triphosphatase (GTPase) Arl8b recruits kinesin-1 and Vps41 (HOPS) onto endosomes carrying misfolded mutant PrP to promote their axonal entry and homotypic fusion toward aggregation inside enlarged endomembranes that we call endoggresomes. This axonal rapid endosomal sorting and transport-dependent aggregation (ARESTA) mechanism forms pathologic PrP endoggresomes that impair calcium dynamics and reduce neuronal viability. Inhibiting ARESTA diminishes endoggresome formation, rescues calcium influx, and prevents neuronal death. Our results identify ARESTA as a key pathway for the regulation of endoggresome formation and a new actionable antiaggregation target to ameliorate neuronal dysfunction in the prionopathies.

13.
Curr Biol ; 31(15): 3440-3449.e7, 2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34146484

RESUMO

Macroautophagy (hereafter referred to as autophagy) is a conserved process that promotes cellular homeostasis through the degradation of cytosolic components, also known as cargo. During autophagy, cargo is sequestered into double-membrane vesicles called autophagosomes, which are predominantly transported in the retrograde direction to the perinuclear region to fuse with lysosomes, thus ensuring cargo degradation.1 The mechanisms regulating directional autophagosomal transport remain unclear. The ATG8 family of proteins associates with autophagosome membranes2 and plays key roles in autophagy, including the movement of autophagosomes. This is achieved via the association of ATG8 with adaptor proteins like FYCO1, involved in the anterograde transport of autophagosomes toward the cell periphery.1,3-5 We previously reported that phosphorylation of LC3B/ATG8 on threonine 50 (LC3B-T50) by the Hippo kinase STK4/MST1 is required for autophagy through unknown mechanisms.6 Here, we show that STK4-mediated phosphorylation of LC3B-T50 reduces the binding of FYCO1 to LC3B. In turn, impairment of LC3B-T50 phosphorylation decreases starvation-induced perinuclear positioning of autophagosomes as well as their colocalization with lysosomes. Moreover, a significantly higher number of LC3B-T50A-positive autophagosomes undergo aberrant anterograde movement to axonal tips in mammalian neurons and toward the periphery of mammalian cells. Our data support a role of a nutrient-sensitive STK4-LC3B-FYCO1 axis in the regulation of the directional transport of autophagosomes, a key step of the autophagy process, via the post-translational modification of LC3B.


Assuntos
Autofagossomos , Proteínas Associadas aos Microtúbulos , Processamento de Proteína Pós-Traducional , Animais , Autofagossomos/metabolismo , Autofagia , Família da Proteína 8 Relacionada à Autofagia/genética , Família da Proteína 8 Relacionada à Autofagia/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Fosforilação
14.
J Mol Neurosci ; 34(1): 9-15, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18157653

RESUMO

The cellular prion protein (PrP(C)) is found prominently at the synapse. However, its role at the nerve termini and elsewhere is unknown. Here we discuss research presented at the 2005 International Institute for Complex Adaptive Matter (I2CAM) first Annual Amyloid Conference that provides insight into the role of synaptic PrP(C). The prion protein can interact and facilitate copper uptake at the synapse, is presumed to oligodimerize to facilitate putative cell-cell adhesion, and it transports toward the synapse by fast microtubule-based anterograde transport. While PrP(C) appears to be involved in all these processes, the mechanisms of PrP(C) function in each of them remain unclear. A role for PrP(C) in these distinct processes suggests a complex role for this protein at the synapse. Unraveling PrP(C) function will likely entail employing combined approaches that take into account its possible multifaceted functions.


Assuntos
Sistema Nervoso/embriologia , Sistema Nervoso/metabolismo , Plasticidade Neuronal/fisiologia , Proteínas PrPC/metabolismo , Sinapses/metabolismo , Animais , Transporte Axonal/fisiologia , Adesão Celular/fisiologia , Cobre/metabolismo , Humanos , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Sistema Nervoso/citologia , Doenças Priônicas/metabolismo , Doenças Priônicas/fisiopatologia , Sinapses/ultraestrutura
15.
Mol Biol Cell ; 16(3): 1056-70, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15616189

RESUMO

During mitosis, chromosome segregation is regulated by a spindle checkpoint mechanism. This checkpoint delays anaphase until all kinetochores are captured by microtubules from both spindle poles, chromosomes congress to the metaphase plate, and the tension between kinetochores and their attached microtubules is properly sensed. Although the spindle checkpoint can be activated in many different cell types, the role of this regulatory mechanism in rapidly dividing embryonic animal cells has remained controversial. Here, using time-lapse imaging of live embryonic cells, we show that chemical or mutational disruption of the mitotic spindle in early Caenorhabditis elegans embryos delays progression through mitosis. By reducing the function of conserved checkpoint genes in mutant embryos with defective mitotic spindles, we show that these delays require the spindle checkpoint. In the absence of a functional checkpoint, more severe defects in chromosome segregation are observed in mutants with abnormal mitotic spindles. We also show that the conserved kinesin CeMCAK, the CENP-F-related proteins HCP-1 and HCP-2, and the core kinetochore protein CeCENP-C all are required for this checkpoint. Our analysis indicates that spindle checkpoint mechanisms are functional in the rapidly dividing cells of an early animal embryo and that this checkpoint can prevent chromosome segregation defects during mitosis.


Assuntos
Caenorhabditis elegans/embriologia , Cromossomos/ultraestrutura , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fuso Acromático , Alelos , Anáfase , Animais , Proteínas de Caenorhabditis elegans/fisiologia , Proteínas Cromossômicas não Histona/fisiologia , Clonagem Molecular , Imuno-Histoquímica , Cinesinas/fisiologia , Cinetocoros/metabolismo , Mitose , Modelos Biológicos , Modelos Genéticos , Nocodazol/farmacologia , Fenótipo , Interferência de RNA , Fatores de Tempo
16.
Elife ; 4: e08833, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26623667

RESUMO

Longevity mechanisms increase lifespan by counteracting the effects of aging. However, whether longevity mechanisms counteract the effects of aging continually throughout life, or whether they act during specific periods of life, preventing changes that precede mortality is unclear. Here, we uncover transcriptional drift, a phenomenon that describes how aging causes genes within functional groups to change expression in opposing directions. These changes cause a transcriptome-wide loss in mRNA stoichiometry and loss of co-expression patterns in aging animals, as compared to young adults. Using Caenorhabditis elegans as a model, we show that extending lifespan by inhibiting serotonergic signals by the antidepressant mianserin attenuates transcriptional drift, allowing the preservation of a younger transcriptome into an older age. Our data are consistent with a model in which inhibition of serotonergic signals slows age-dependent physiological decline and the associated rise in mortality levels exclusively in young adults, thereby postponing the onset of major mortality.


Assuntos
Envelhecimento , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Longevidade/efeitos dos fármacos , Antagonistas da Serotonina/administração & dosagem , Transcrição Gênica , Animais , Perfilação da Expressão Gênica , Mianserina/administração & dosagem
17.
Nat Rev Drug Discov ; 14(11): 759-80, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26338154

RESUMO

The aggregation of specific proteins is hypothesized to underlie several degenerative diseases, which are collectively known as amyloid disorders. However, the mechanistic connection between the process of protein aggregation and tissue degeneration is not yet fully understood. Here, we review current and emerging strategies to ameliorate aggregation-associated degenerative disorders, with a focus on disease-modifying strategies that prevent the formation of and/or eliminate protein aggregates. Persuasive pharmacological and genetic evidence now supports protein aggregation as the cause of postmitotic tissue dysfunction or loss. However, a more detailed understanding of the factors that trigger and sustain aggregate formation and of the structure-activity relationships underlying proteotoxicity is needed to develop future disease-modifying therapies.


Assuntos
Doenças Neurodegenerativas/tratamento farmacológico , Agregação Patológica de Proteínas/tratamento farmacológico , Transporte Proteico/efeitos dos fármacos , Animais , Fatores Biológicos/farmacologia , Fatores Biológicos/uso terapêutico , Humanos , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Pré-Albumina/antagonistas & inibidores , Pré-Albumina/química , Pré-Albumina/metabolismo , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Dobramento de Proteína/efeitos dos fármacos , Relação Estrutura-Atividade , Resultado do Tratamento
18.
Annu Rev Biophys ; 43: 141-69, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24702007

RESUMO

Axonal transport is indispensable for the distribution of vesicles, organelles, messenger RNAs (mRNAs), and signaling molecules along the axon. This process is mediated by kinesins and dyneins, molecular motors that bind to cargoes and translocate on microtubule tracks. Tight modulation of motor protein activity is necessary, but little is known about the molecules and mechanisms that regulate transport. Moreover, evidence suggests that transport impairments contribute to the initiation or progression of neurodegenerative diseases, or both, but the mechanisms by which motor activity is affected in disease are unclear. In this review, we discuss some of the physical and biophysical properties that influence motor regulation in healthy neurons. We further discuss the evidence for the role of transport in neurodegeneration, highlighting two pathways that may contribute to transport impairment-dependent disease: genetic mutations or variation, and protein aggregation. Understanding how and when transport parameters change in disease will help delineate molecular mechanisms of neurodegeneration.


Assuntos
Transporte Axonal , Proteínas Motores Moleculares/metabolismo , Neurônios/metabolismo , Animais , Humanos , Doenças Neurodegenerativas/metabolismo
19.
J Vis Exp ; (92): e52029, 2014 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-25406537

RESUMO

Understanding the mechanisms by which molecular motors coordinate their activities to transport vesicular cargoes within neurons requires the quantitative analysis of motor/cargo associations at the single vesicle level. The goal of this protocol is to use quantitative fluorescence microscopy to correlate ("map") the position and directionality of movement of live cargo to the composition and relative amounts of motors associated with the same cargo. "Cargo mapping" consists of live imaging of fluorescently labeled cargoes moving in axons cultured on microfluidic devices, followed by chemical fixation during recording of live movement, and subsequent immunofluorescence (IF) staining of the exact same axonal regions with antibodies against motors. Colocalization between cargoes and their associated motors is assessed by assigning sub-pixel position coordinates to motor and cargo channels, by fitting Gaussian functions to the diffraction-limited point spread functions representing individual fluorescent point sources. Fixed cargo and motor images are subsequently superimposed to plots of cargo movement, to "map" them to their tracked trajectories. The strength of this protocol is the combination of live and IF data to record both the transport of vesicular cargoes in live cells and to determine the motors associated to these exact same vesicles. This technique overcomes previous challenges that use biochemical methods to determine the average motor composition of purified heterogeneous bulk vesicle populations, as these methods do not reveal compositions on single moving cargoes. Furthermore, this protocol can be adapted for the analysis of other transport and/or trafficking pathways in other cell types to correlate the movement of individual intracellular structures with their protein composition. Limitations of this protocol are the relatively low throughput due to low transfection efficiencies of cultured primary neurons and a limited field of view available for high-resolution imaging. Future applications could include methods to increase the number of neurons expressing fluorescently labeled cargoes.


Assuntos
Transporte Axonal/fisiologia , Axônios/metabolismo , Proteínas Motores Moleculares/metabolismo , Neurônios/metabolismo , Vesículas Transportadoras/metabolismo , Animais , Hipocampo/citologia , Camundongos , Técnicas Analíticas Microfluídicas/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA